Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Neurosci ; 44(11)2024 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-38316563

RÉSUMÉ

Cooling sensations arise inside the mouth during ingestive and homeostasis behaviors. Oral presence of cooling temperature engages the cold and menthol receptor TRPM8 (transient receptor potential melastatin 8) on trigeminal afferents. Yet, how TRPM8 influences brain and behavioral responses to oral temperature is undefined. Here we used in vivo neurophysiology to record action potentials stimulated by cooling and warming of oral tissues from trigeminal nucleus caudalis neurons in female and male wild-type and TRPM8 gene deficient mice. Using these lines, we also measured orobehavioral licking responses to cool and warm water in a novel, temperature-controlled fluid choice test. Capture of antidromic electrophysiological responses to thalamic stimulation identified that wild-type central trigeminal neurons showed diverse responses to oral cooling. Some neurons displayed relatively strong excitation to cold <10°C (COLD neurons) while others responded to only a segment of mild cool temperatures below 30°C (COOL neurons). Notably, TRPM8 deficient mice retained COLD-type but lacked COOL cells. This deficit impaired population responses to mild cooling temperatures below 30°C and allowed warmth-like (≥35°C) neural activity to pervade the normally innocuous cool temperature range, predicting TRPM8 deficient mice would show anomalously similar orobehavioral responses to warm and cool temperatures. Accordingly, TRPM8 deficient mice avoided both warm (35°C) and mild cool (≤30°C) water and sought colder temperatures in fluid licking tests, whereas control mice avoided warm but were indifferent to mild cool and colder water. Results imply TRPM8 input separates cool from warm temperature sensing and suggest other thermoreceptors also participate in oral cooling sensation.


Sujet(s)
Canaux cationiques TRPM , Souris , Mâle , Animaux , Femelle , Canaux cationiques TRPM/génétique , Basse température , Neurones , Température , Thermoception/physiologie , Eau
3.
Front Neurosci ; 17: 1267542, 2023.
Article de Anglais | MEDLINE | ID: mdl-38033546

RÉSUMÉ

Developmental exposure to ethanol is a leading cause of cognitive, emotional and behavioral problems, with fetal alcohol spectrum disorder (FASD) affecting more than 1:100 children. Recently, comorbid sleep deficits have been highlighted in these disorders, with sleep repair a potential therapeutic target. Animal models of FASD have shown non-REM (NREM) sleep fragmentation and slow-wave oscillation impairments that predict cognitive performance. Here we use a mouse model of perinatal ethanol exposure to explore whether reduced sleep pressure may contribute to impaired NREM sleep, and compare the function of a brain network reported to be impacted by insomnia-the Salience network-in developmental ethanol-exposed mice with sleep-deprived, saline controls. Mice were exposed to ethanol or saline on postnatal day 7 (P7) and allowed to mature to adulthood for testing. At P90, telemetered cortical recordings were made for assessment of NREM sleep in home cage before and after 4 h of sleep deprivation to assess basal NREM sleep and homeostatic NREM sleep response. To assess Salience network functional connectivity, mice were exposed to the 4 h sleep deprivation period or left alone, then immediately sacrificed for immunohistochemical analysis of c-Fos expression. The results show that developmental ethanol severely impairs both normal rebound NREM sleep and sleep deprivation induced increases in slow-wave activity, consistent with reduced sleep pressure. Furthermore, the Salience network connectome in rested, ethanol-exposed mice was most similar to that of sleep-deprived, saline control mice, suggesting a sleep deprivation-like state of Salience network function after developmental ethanol even without sleep deprivation.

4.
J Neurosci ; 42(9): 1719-1737, 2022 03 02.
Article de Anglais | MEDLINE | ID: mdl-35027408

RÉSUMÉ

Trigeminal neurons convey somatosensory information from craniofacial tissues. In mouse brain, ascending projections from medullary trigeminal neurons arrive at taste neurons in the parabrachial (PB) nucleus, suggesting that taste neurons participate in somatosensory processing. However, the cell types that support this convergence were undefined. Using Cre-directed optogenetics and in vivo neurophysiology in anesthetized mice of both sexes, here we studied whether transient receptor potential vanilloid 1 (TRPV1)-lineage nociceptive and thermosensory fibers are primary neurons that drive trigeminal circuits reaching PB taste cells. We monitored spiking activity in individual PB neurons during photoexcitation of the terminals of TRPV1-lineage fibers arriving at the dorsal trigeminal nucleus caudalis, which relays orofacial somatosensory messages to the PB area. We also recorded PB neural responses to oral delivery of taste, chemesthetic, and thermal stimuli. We found that optical excitation of TRPV1-lineage fibers elicited responses in traditionally defined taste neurons in lateral PB nuclei. The tuning of neurons across diverse tastes associated with their sensitivity to TRPV1-lineage fiber stimulation, which only sparingly engaged neurons oriented to preferred tastes like sucrose. Moreover, neurons responsive to photostimulation of TRPV1-lineage afferents showed strong responses to temperature including noxious heat, which predominantly excited PB bitter taste cells. Multivariate and machine learning analyses revealed the PB confluence of TRPV1-lineage signals with taste captured sensory valence information shared across aversive gustatory, nociceptive, and thermal stimuli. Our results reveal that TRPV1-lineage fibers, which have defined roles in thermosensation and pain, communicate with PB taste neurons. This multisensory convergence supports dependencies between gustatory and somatosensory hedonic representations in the brain.SIGNIFICANCE STATEMENT The parabrachial (PB) nucleus participates in autonomic and integrative neural processing for diverse sensory modalities. We recently found in mice that trigeminal neurons supplying craniofacial somatosensation project to PB neurons sensitive to tastes. Here, we show that trigeminal projections to PB gustatory cells are driven by a genetic class of thermosensory and nociceptive fiber. Input from these fibers was associated with PB neural sensitivity to aversive oral temperatures and tastes and supported a multimodal neural representation of sensory valence across gustatory, nociceptive, and thermal stimuli. These results reveal gustation and somatosensation to be only components of a larger PB code that captures sensory value. Defining this circuit has implications for understanding the neural representation of taste, temperature, and also pain-related phenomena.


Sujet(s)
Noyau parabrachial , Animaux , Femelle , Mâle , Moelle allongée/physiologie , Souris , Neurones/physiologie , Douleur , Noyau parabrachial/physiologie , Canaux cationiques TRPV , Goût/physiologie
5.
Curr Opin Physiol ; 20: 16-22, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-33937598

RÉSUMÉ

Temperature sensation contributes to human enjoyment of foods and beverages. The mouthfeel of warmed foods or drinking ice-cold water on a hot day are respectively pleasant and refreshing. Although historically under-studied for a role in food preference, new data have shed light on how oral temperature sensing and thermoreceptor mechanisms inside the mouth influence ingestive acceptance behaviors in rodent models used in flavor neurobiology. Moreover, recent functional data have uncovered a broad diversity of thermosensory neurons in primary afferents and brain pathways that signal oral temperature. This review will discuss some of the progress made in these areas. Ultimately, unraveling the biological basis of oral temperature sensing will be critical to reveal how thermosensory factors interact with other orosensory modalities to shape ingestive preferences. Elucidating oral thermal processing will also be key for establishing general principles of temperature coding by the nervous system.

6.
eNeuro ; 6(6)2019.
Article de Anglais | MEDLINE | ID: mdl-31624176

RÉSUMÉ

The flavoring agent menthol elicits complex orosensory and behavioral effects including perceived cooling at low concentrations and irritation and ingestive avoidance at higher intensities. Oral menthol engages the cold-activated transient receptor potential (TRP) ion channel TRP melastatin 8 (TRPM8) on trigeminal fibers, although its aversive feature was discussed to involve activation of TRP ankyrin 1 (TRPA1) associated with nociceptive processing. Here, we studied the roles of TRPM8 and TRPA1 in orosensory responding to menthol by subjecting mice gene deficient for either channel to brief-access exposure tests, which measure immediate licking responses to fluid stimuli to capture sensory/tongue control of behavior. Stimuli included aqueous concentration series of (-)-menthol [0 (water), 0.3, 0.5, 0.7, 1.0, 1.5, and 2.3 mM] and the aversive bitter taste stimulus quinine-HCl (0, 0.01, 0.03, 0.1, 0.3, 1, and 3 mM). Concentration-response data were generated from daily brief-access tests conducted in lickometers, which recorded the number of licks water-restricted mice emitted to a randomly selected stimulus concentration over a block of several 10-s stimulus presentations. Wild-type mice showed aversive orosensory responses to menthol above 0.7 mM. Oral aversion to menthol was reduced in mice deficient for TRPA1 but not TRPM8. Oral aversion to quinine was similar between TRPA1 mutant and control mice but stronger than avoidance of menthol. This implied menthol avoidance under the present conditions represented a moderate form of oral aversion. These data reveal TRPA1 contributes to the oral sensory valence of menthol and have implications for how input from TRPA1 and TRPM8 shapes somatosensory-guided behaviors.


Sujet(s)
Apprentissage par évitement/physiologie , Menthol/administration et posologie , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire/métabolisme , Animaux , Apprentissage par évitement/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Comportement animal/physiologie , Souris , Souris knockout , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire/génétique , Canaux cationiques TRPM/génétique , Canaux cationiques TRPM/métabolisme
7.
J Neurosci ; 39(9): 1631-1648, 2019 02 27.
Article de Anglais | MEDLINE | ID: mdl-30606758

RÉSUMÉ

Taste and somatosensation both mediate protective behaviors. Bitter taste guides avoidance of ingestion of toxins while pain sensations, such as noxious heat, signal adverse conditions to ward off harm. Although brain pathways for taste and somatosensation are typically studied independently, prior data suggest that they intersect, potentially reflecting their common protective role. To investigate this, we applied electrophysiologic and optogenetic techniques in anesthetized mice of both sexes to evaluate relationships between oral somatosensory and taste activity in the parabrachial nucleus (PbN), implicated for roles in gustation and pain. Spikes were recorded from taste-active PbN neurons tested with oral delivery of thermal and chemesthetic stimuli, including agonists of nocisensitive transient receptor potential (TRP) ion channels on somatosensory fibers. Gustatory neurons were also tested to follow electrical pulse stimulation of an oral somatosensory region of the spinal trigeminal subnucleus caudalis (Vc), which projects to the PbN. Neurons composed classic taste groups, including sodium, electrolyte, appetitive, or bitter cells. Across groups, most neurons spiked to Vc pulse stimulation, implying that trigeminal projections reach PbN gustatory neurons. Among such cells, a subpopulation responsive to the bitter taste stimuli quinine and cycloheximide, and aversive concentrations of sodium, cofired to agonists of nocisensitive TRP channels, including capsaicin, mustard oil, and noxious heat. Such neurons populated the lateral PbN. Further, nociceptive activity in PbN bitter taste neurons was suppressed during optogenetic-assisted inhibition of the Vc, implying convergent trigeminal input contributed to such activity. Our results reveal a novel role for PbN gustatory cells in cross-system signaling related to protection.SIGNIFICANCE STATEMENT Prior data suggest that gustatory and trigeminal neural pathways intersect and overlap in the parabrachial area. However, no study has directly examined such overlap and why it may exist. Here we found that parabrachial gustatory neurons can receive afferent projections from trigeminal nuclei and fire to oral nociceptive stimuli that excite somatosensory receptors and fibers. Activation to aversive nociceptive stimuli in gustatory cells was associated with responding to behaviorally avoided bitter tastants. We were further able to show that silencing trigeminal projections inhibited nociceptive activity in parabrachial bitter taste neurons. Our results imply that in the parabrachial area, there is predictable overlap between taste and somatosensory processing related to protective coding and that classically defined taste neurons contribute to this process.


Sujet(s)
Nociception , Noyau parabrachial/physiologie , Cellules réceptrices sensorielles/métabolisme , Perception du goût , Potentiels d'action , Animaux , Capsaïcine/pharmacologie , Cycloheximide/pharmacologie , Femelle , Mâle , Souris , Souris de lignée C57BL , Moutarde (plante) , Noyau parabrachial/cytologie , Huiles végétales/pharmacologie , Quinine/pharmacologie , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Cellules réceptrices sensorielles/physiologie , Goût , Canaux cationiques TRP/métabolisme
8.
Am J Physiol Regul Integr Comp Physiol ; 313(4): R305-R321, 2017 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-28794101

RÉSUMÉ

Taste stimuli have a temperature that can stimulate thermosensitive neural machinery in the mouth during gustatory experience. Although taste and oral temperature are sometimes discussed as different oral sensory modalities, there is a body of literature that demonstrates temperature is an important component and modulator of the intensity of gustatory neural and perceptual responses. Available data indicate that the influence of temperature on taste, herein referred to as "thermogustation," can vary across taste qualities, can also vary among stimuli presumed to share a common taste quality, and is conditioned on taste stimulus concentration, with neuronal and psychophysical data revealing larger modulatory effects of temperature on gustatory responding to weakened taste solutions compared with concentrated. What is more, thermogustation is evidenced to involve interplay between mouth and stimulus temperature. Given these and other dependencies, identifying principles by which thermal input affects gustatory information flow in the nervous system may be important for ultimately unravelling the organization of neural circuits for taste and defining their involvement with multisensory processing related to flavor. Yet thermal effects are relatively understudied in gustatory neuroscience. Major gaps in our understanding of the mechanisms and consequences of thermogustation include delineating supporting receptors, the potential involvement of oral thermal and somatosensory trigeminal neurons in thermogustatory interactions, and the broader operational roles of temperature in gustatory processing. This review will discuss these and other issues in the context of the literature relevant to understanding thermogustation.


Sujet(s)
Régulation de la température corporelle/physiologie , Modèles neurologiques , Bouche/physiologie , Cellules réceptrices sensorielles/physiologie , Goût/physiologie , Thermoception/physiologie , Animaux , Température du corps/physiologie , Humains
9.
Chem Senses ; 41(5): 457-71, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26976122

RÉSUMÉ

Oral temperature is a component and modifier of taste perception. Both trigeminal (V) and taste-sensitive cells, including those in the nucleus of the solitary tract (NTS), can respond to oral temperature. However, functional associations in thermal sensitivity between V and gustatory neurons are poorly understood. To study this we recorded electrophysiological responses to oral stimulation with cool (9, 15, 25, 32, and 34 °C) and warm (40 and 45 °C) temperatures from medullary V (n = 45) and taste-sensitive NTS (n = 27) neurons in anesthetized mice. Results showed temperatures below 34 °C activated the majority of V neurons but only a minority of NTS units. V neurons displayed larger responses to cooling and responded to temperatures that poorly stimulated NTS cells. Multivariate analyses revealed different temperatures induced larger differences in responses across V compared with NTS neurons, indicating V pathways possess greater capacity to signal temperature. Conversely, responses to temperature in NTS units associated with gustatory tuning. Further analyses identified two types of cooling-sensitive V neurons oriented toward innocuous or noxious cooling. Multivariate analyses indicated the combined response of these cells afforded distinction among a broad range of cool temperatures, suggesting multiple types of V neurons work together to represent oral cooling.


Sujet(s)
Neurones/physiologie , Perception du goût/physiologie , Animaux , Stimulation électrique , Potentiels évoqués , Femelle , Mâle , Souris , Souris de lignée C3H , Analyse multifactorielle , Noyau du tractus solitaire/physiologie , Température
10.
Chemosens Percept ; 8(2): 46-52, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26388965

RÉSUMÉ

INTRODUCTION: This mini-review discusses some of the parallels between rodent neurophysiological and human psychophysical data concerning temperature effects on sweet taste. METHODS AND PURPOSE: "Sweet" is an innately rewarding taste sensation that is associated in part with foods that contain calories in the form of sugars. Humans and other mammals can show unconditioned preference for select sweet stimuli. Such preference is poised to influence diet selection and, in turn, nutritional status, which underscores the importance of delineating the physiological mechanisms for sweet taste with respect to their influence on human health. Advances in our knowledge of the biology of sweet taste in humans have arisen in part through studies on mechanisms of gustatory processing in rodent models. Along this line, recent work has revealed there are operational parallels in neural systems for sweet taste between mice and humans, as indexed by similarities in the effects of temperature on central neurophysiological and psychophysical responses to sucrose in these species. Such association strengthens the postulate that rodents can serve as effective models of particular mechanisms of appetitive taste processing. Data supporting this link are discussed here, as are rodent and human data that shed light on relationships between mechanisms for sweet taste and ingestive disorders, such as alcohol abuse. RESULTS AND CONCLUSIONS: Rodent models have utility for understanding mechanisms of taste processing that may pertain to human flavor perception. Importantly, there are limitations to generalizing data from rodents, albeit parallels across species do exist.

11.
J Neurophysiol ; 113(7): 2700-12, 2015 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-25673737

RÉSUMÉ

The temperature of taste stimuli can modulate gustatory processing. Perceptual data indicate that the adapted temperature of oral epithelia also influences gustation, although little is known about the neural basis of this effect. Here, we electrophysiologically recorded orosensory responses (spikes) to 25°C (cool) and 35°C (warm) solutions of sucrose (0.1 and 0.3 M), NaCl (0.004, 0.1, and 0.3 M), and water from taste-sensitive neurons in the nucleus of the solitary tract in mice under varied thermal adaptation of oral epithelia. Conditions included presentation of taste stimuli isothermal to adaptation temperatures of 25°C (constant cooling) and 35°C (constant warming), delivery of 25°C stimuli following 35°C adaptation (relative cooling), and presentation of 35°C stimuli following 25°C adaptation (relative warming). Responses to sucrose in sucrose-oriented cells (n = 15) were enhanced under the constant and relative warming conditions compared with constant cooling, where contiguous cooling across adaptation and stimulus periods induced the lowest and longest latency responses to sucrose. Yet compared with constant warming, cooling sucrose following warm adaptation (relative cooling) only marginally reduced activity to 0.1 M sucrose and did not alter responses to 0.3 M sucrose. Thus, warmth adaptation counteracted the attenuation in sucrose activity associated with stimulus cooling. Analysis of sodium-oriented (n = 25) neurons revealed adaptation to cool water, and cooling taste solutions enhanced unit firing to 0.004 M (perithreshold) NaCl, whereas warmth adaptation and stimulus warming could facilitate activity to 0.3 M NaCl. The concentration dependence of this thermal effect may reflect a dual effect of temperature on the sodium reception mechanism that drives sodium-oriented cells.


Sujet(s)
Potentiels d'action/physiologie , Température du corps/physiologie , Moelle allongée/physiologie , Muqueuse de la bouche/physiologie , Calicules gustatifs/physiologie , Goût/physiologie , Adaptation physiologique/physiologie , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Lignées consanguines de souris , Stimulation physique/méthodes , Température
12.
J Neurophysiol ; 112(7): 1667-77, 2014 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-24966301

RÉSUMÉ

Temperature can modify neural and behavioral responses to taste stimuli that elicit "sweetness," a perception linked to intake of calorie-laden foods. However, the role of temperature in the neural representation of sweet taste is poorly understood. Here we made electrophysiological recordings from gustatory neurons in the medulla of inbred mice to study how adjustments in taste solution temperature to cool (18°C), ambient (22°C), and warm (30°C and 37°C) values changed the magnitude and latency of gustatory activity to sucrose (0, 0.05, 0.1, 0.17, 0.31, and 0.56 M). Analysis of 22 sucrose-best neurons revealed that temperature markedly influenced responses to sucrose, which, across concentrations, were largest when solutions were warmed to 30°C. However, reducing solution temperature from warm to ambient to cool progressively steepened the slope of the sucrose concentration-response function computed across cells (P < 0.05), indicating that mean activity to sucrose increased more rapidly with concentration steps under cooling than with warming. Thus the slope of the sucrose concentration-response function shows an inverse relation with temperature. Temperature also influenced latency to the first spike of the sucrose response. Across neurons, latencies were shorter when sucrose solutions were warmed and longer, by hundreds of milliseconds, when solutions were cooled (P < 0.05), indicating that temperature is also a temporal parameter of sucrose activity. Our findings reveal that temperature systematically modifies the timing of gustatory activity to sucrose in the mammalian brain and how this activity changes with concentration. Results further highlight how oral somatosensory cues function as physiological modulators of gustatory processing.


Sujet(s)
Moelle allongée/physiologie , Neurones/physiologie , Perception du goût/physiologie , Potentiels d'action , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Saccharose , Température
13.
J Neurophysiol ; 110(5): 1117-29, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23761701

RÉSUMÉ

Changes in oral temperature can influence taste perception, indicating overlap among mechanisms for taste and oral somesthesis. Medullary gustatory neurons can show cosensitivity to temperature, albeit how these cells process combined taste and thermal input is poorly understood. Here, we electrophysiologically recorded orosensory responses (spikes) from 39 taste-sensitive neurons in the nucleus tractus solitarii of anesthetized mice during oral delivery of tastants adjusted to innocuous cool (16 and 18°C), room (22°C, baseline), and warm (30 and 37°C) oral temperatures. Stimuli included (in mM) 100 sucrose, 30 NaCl, 3 HCl, 3 quinine, an umami mixture, and water. Although cooled water excited few cells, water warmed to 30 and 37°C significantly excited 33% and 64% of neurons, respectively. Warmth induced responses of comparable magnitude to room temperature tastants. Furthermore, warming taste solutions influenced the distribution of gustatory responses among neurons and increased (P < 0.05) neuronal breadth of tuning across taste qualities. The influence of warmth on response magnitude was stimulus specific. Across neurons, warming facilitated responses to sucrose and umami in a superadditive manner, as these responses exceeded (P < 0.05) the arithmetic sum of activity to warming alone and the taste stimulus tested at room temperature. Superadditive increases (P < 0.05) in responding were also noted in some cells for warmed HCl. Yet warming induced only simple additive or subtractive effects on responses to quinine and NaCl. Data show temperature is a parameter of gustatory processing, like taste quality and concentration, in medullary circuits for taste.


Sujet(s)
Température du corps , Neurones/physiologie , Noyau du tractus solitaire/physiologie , Goût/physiologie , Animaux , Femelle , Mâle , Souris , Souris de lignée C3H
14.
PLoS One ; 7(7): e41597, 2012.
Article de Anglais | MEDLINE | ID: mdl-22844505

RÉSUMÉ

A growing literature suggests taste stimuli commonly classified as "bitter" induce heterogeneous neural and perceptual responses. Here, the central processing of bitter stimuli was studied in mice with genetically controlled bitter taste profiles. Using these mice removed genetic heterogeneity as a factor influencing gustatory neural codes for bitter stimuli. Electrophysiological activity (spikes) was recorded from single neurons in the nucleus tractus solitarius during oral delivery of taste solutions (26 total), including concentration series of the bitter tastants quinine, denatonium benzoate, cycloheximide, and sucrose octaacetate (SOA), presented to the whole mouth for 5 s. Seventy-nine neurons were sampled; in many cases multiple cells (2 to 5) were recorded from a mouse. Results showed bitter stimuli induced variable gustatory activity. For example, although some neurons responded robustly to quinine and cycloheximide, others displayed concentration-dependent activity (p<0.05) to quinine but not cycloheximide. Differential activity to bitter stimuli was observed across multiple neurons recorded from one animal in several mice. Across all cells, quinine and denatonium induced correlated spatial responses that differed (p<0.05) from those to cycloheximide and SOA. Modeling spatiotemporal neural ensemble activity revealed responses to quinine/denatonium and cycloheximide/SOA diverged during only an early, at least 1 s wide period of the taste response. Our findings highlight how temporal features of sensory processing contribute differences among bitter taste codes and build on data suggesting heterogeneity among "bitter" stimuli, data that challenge a strict monoguesia model for the bitter quality.


Sujet(s)
Phénomènes électrophysiologiques , Noyau du tractus solitaire/physiologie , Perception du goût/physiologie , Animaux , Femelle , Mâle , Souris , Neurones/cytologie , Stimulation physique , Noyau du tractus solitaire/cytologie , Analyse spatio-temporelle
15.
Addict Biol ; 17(2): 423-36, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22129513

RÉSUMÉ

Alcohol activates orosensory circuits that project to motivationally relevant limbic forebrain areas that control appetite, feeding and drinking. To date, limited data exists regarding the contribution of chemosensory-derived ethanol reinforcement to ethanol preference and consumption. Measures of taste reactivity to intra-orally infused ethanol have not found differences in initial orofacial responses to alcohol between alcohol-preferring (P) and alcohol-non-preferring (NP) genetically selected rat lines. Yet, in voluntary intake tests, P rats prefer highly concentrated ethanol upon initial exposure, suggesting an early sensory-mediated attraction. Here, we directly compared self-initiated chemosensory responding for alcohol and prototypic sweet, bitter and oral trigeminal stimuli among selectively bred P, NP and non-selected Wistar (WI) outbred lines to determine whether differential sensory responsiveness to ethanol and its putative sensory components are phenotypically associated with genetically influenced alcohol preference. Rats were tested for immediate short-term lick responses to alcohol (3-40%), sucrose (0.01-1 M), quinine (0.01-3 mM) and capsaicin (0.003-1 mM) in a brief-access assay designed to index orosensory-guided behavior. P rats exhibited elevated short-term lick responses to both alcohol and sucrose relative to NP and WI lines across a broad range of concentrations of each stimulus and in the absence of blood alcohol levels that would produce significant post-absorptive effects. There was no consistent relationship between genetically mediated alcohol preference and orosensory avoidance of quinine or capsaicin. These data indicate that enhanced initial chemosensory attraction to ethanol and sweet stimuli are phenotypes associated with genetic alcohol preference and are considered within the framework of downstream activation of oral appetitive reward circuits.


Sujet(s)
Consommation d'alcool/génétique , Éthanol/pharmacologie , Motivation/génétique , Goût/génétique , Animaux , Appétit/effets des médicaments et des substances chimiques , Appétit/génétique , Capsaïcine/pharmacologie , Conditionnement opérant , Éthanol/métabolisme , Hétérogénéité génétique , Mâle , Quinine/pharmacologie , Rats , , Récompense , Autoadministration , Agents du système nerveux sensoriel/pharmacologie , Saccharose/pharmacologie , Édulcorants/pharmacologie
16.
J Neurophysiol ; 106(6): 3145-56, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21918002

RÉSUMÉ

In randomly bred rats, orally applied ethanol stimulates neural substrates for appetitive sweet taste. To study associations between ethanol's oral sensory characteristics and genetically mediated ethanol preference, we made electrophysiological recordings of oral responses (spike density) by taste-sensitive nucleus tractus solitarii neurons in anesthetized selectively bred ethanol-preferring (P) rats and their genetically heterogeneous Wistar (W) control strain. Stimuli (25 total) included ethanol [3%, 5%, 10%, 15%, 25%, and 40% (vol/vol)], a sucrose series (0.01, 0.03, 0.1, 0.3, 0.5, and 1 M), and other sweet, salt, acidic, and bitter stimuli; 50 P and 39 W neurons were sampled. k-means clustering applied to the sucrose response series identified cells showing high (S(1)) or relatively low (S(0)) sensitivity to sucrose. A three-way factorial analysis revealed that activity to ethanol was influenced by a neuron's sensitivity to sucrose, ethanol concentration, and rat line (P = 0.01). Ethanol produced concentration-dependent responses in S(1) neurons that were larger than those in S(0) cells. Although responses to ethanol by S(1) cells did not differ between lines, neuronal firing rates to ethanol in S(0) cells increased across concentration only in P rats. Correlation and multivariate analyses revealed that ethanol evoked responses in W neurons that were strongly and selectively associated with activity to sweet stimuli, whereas responses to ethanol by P neurons were not easily associated with activity to representative sweet, sodium salt, acidic, or bitter stimuli. These findings show differential central neural representation of oral ethanol between genetically heterogeneous rats and P rats genetically selected to prefer alcohol.


Sujet(s)
Dépresseurs du système nerveux central/administration et posologie , Éthanol/administration et posologie , Préférences alimentaires/physiologie , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Noyau du tractus solitaire/cytologie , Goût/physiologie , Potentiels d'action/effets des médicaments et des substances chimiques , Administration par voie orale , Voies afférentes/physiologie , Animaux , Relation dose-effet des médicaments , Préférences alimentaires/effets des médicaments et des substances chimiques , Mâle , Modèles biologiques , Rats , Rat Wistar , Statistiques comme sujet , Activation chimique , Estomac/innervation , Saccharose/administration et posologie , Édulcorants/administration et posologie
17.
Mo Med ; 107(4): 247-51, 2010.
Article de Anglais | MEDLINE | ID: mdl-20806835

RÉSUMÉ

This paper reviews the physiology of taste processing and ingestive decisions.


Sujet(s)
Comportement du consommateur , Régime alimentaire , Goût/physiologie , Appétit/physiologie , Humains , Conduction nerveuse/physiologie , Calicules gustatifs/physiologie
18.
Physiol Genomics ; 41(3): 232-43, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20145204

RÉSUMÉ

Elevated alcohol consumption is associated with enhanced preference for sweet substances across species and may be mediated by oral alcohol-induced activation of neurobiological substrates for sweet taste. Here, we directly examined the contribution of the T1r3 receptor protein, important for sweet taste detection in mammals, to ethanol intake and preference and the neural processing of ethanol taste by measuring behavioral and central neurophysiological responses to oral alcohol in T1r3 receptor-deficient mice and their C57BL/6J background strain. T1r3 knockout and wild-type mice were tested in behavioral preference assays for long-term voluntary intake of a broad concentration range of ethanol, sucrose, and quinine. For neurophysiological experiments, separate groups of mice of each genotype were anesthetized, and taste responses to ethanol and stimuli of different taste qualities were electrophysiologically recorded from gustatory neurons in the nucleus of the solitary tract. Mice lacking the T1r3 receptor were behaviorally indifferent to alcohol (i.e., ∼50% preference values) at concentrations typically preferred by wild-type mice (5-15%). Central neural taste responses to ethanol in T1r3-deficient mice were significantly lower compared with C57BL/6J controls, a strain for which oral ethanol stimulation produced a concentration-dependent activation of sweet-responsive NTS gustatory neurons. An attenuated difference in ethanol preference between knockouts and controls at concentrations >15% indicated that other sensory and/or postingestive effects of ethanol compete with sweet taste input at high concentrations. As expected, T1r3 knockouts exhibited strongly suppressed behavioral and neural taste responses to sweeteners but did not differ from wild-type mice in responses to prototypic salt, acid, or bitter stimuli. These data implicate the T1r3 receptor in the sensory detection and transduction of ethanol taste.


Sujet(s)
Éthanol/administration et posologie , Éthanol/pharmacologie , Neurones/physiologie , Récepteurs couplés aux protéines G/métabolisme , Goût/effets des médicaments et des substances chimiques , Administration par voie orale , Animaux , Comportement alimentaire/effets des médicaments et des substances chimiques , Femelle , Préférences alimentaires/physiologie , Mâle , Souris de lignée C57BL , Souris knockout , Neurones/effets des médicaments et des substances chimiques , Stimulation physique , Quinine/administration et posologie , Quinine/pharmacologie , Normes de référence , Saccharose/administration et posologie , Saccharose/pharmacologie
19.
J Neurophysiol ; 101(5): 2459-71, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19279151

RÉSUMÉ

T1r3 is a critical subunit of T1r sweet taste receptors. Here we studied how the absence of T1r3 impacts responses to sweet stimuli by taste neurons in the nucleus tractus solitarius (NTS) of the mouse. The consequences bear on the multiplicity of sweet taste receptors and how T1r3 influences the distribution of central gustatory neurons. Taste responses to glycine, sucrose, NaCl, HCl, and quinine were electrophysiologically recorded from single NTS neurons in anesthetized T1r3 knockout (KO) and wild-type (WT) C57BL/6 mice. Other stimuli included l-proline, d-fructose, d-glucose, d-sorbitol, Na-saccharin, acesulfame-K, monosodium glutamate, NaNO(3), Na-acetate, citric acid, KCl, denatonium, and papaverine. Forty-one WT and 41 KO neurons were recorded. Relative to WT, KO responses to all sweet stimuli were significantly lower, although the degree of attenuation differed among stimuli, with near zero responses to sugars but salient residual activity to artificial sweeteners and glycine. Residual KO across-neuron responses to sweet stimuli were variably similar to nonsweet responses, as indexed by multivariate and correlation analyses. In some cases, this suggested that residual KO activity to "sweet" stimuli could be mediated by nonsweet taste receptors, implicating T1r3 receptors as primary contributors to NTS sweet processing. The influence of T1r3 on the distribution of NTS neurons was evaluated by comparing neuron types that emerged between WT and KO cells. Neurons tuned toward sweet stimuli composed 34% of the WT sample but did not appear among KO cells. Input from T1r3-containing receptors critically guides the normal development of NTS neurons oriented toward sweet tastants.


Sujet(s)
Cellules réceptrices sensorielles/physiologie , Noyau du tractus solitaire/cytologie , Récepteur de type Toll-3/physiologie , Potentiels d'action/effets des médicaments et des substances chimiques , Potentiels d'action/génétique , Analyse de variance , Animaux , Relation dose-effet des médicaments , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Statistiques comme sujet , Activation chimique , Édulcorants/pharmacologie , Goût/effets des médicaments et des substances chimiques , Goût/génétique , Seuil du goût/effets des médicaments et des substances chimiques , Seuil du goût/génétique , Récepteur de type Toll-3/déficit
20.
BMC Neurosci ; 8 Suppl 3: S5, 2007 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-17903281

RÉSUMÉ

Although there have been many recent advances in the field of gustatory neurobiology, our knowledge of how the nervous system is organized to process information about taste is still far from complete. Many studies on this topic have focused on understanding how gustatory neural circuits are spatially organized to represent information about taste quality (e.g., "sweet", "salty", "bitter", etc.). Arguments pertaining to this issue have largely centered on whether taste is carried by dedicated neural channels or a pattern of activity across a neural population. But there is now mounting evidence that the timing of neural events may also importantly contribute to the representation of taste. In this review, we attempt to summarize recent findings in the field that pertain to these issues. Both space and time are variables likely related to the mechanism of the gustatory neural code: information about taste appears to reside in spatial and temporal patterns of activation in gustatory neurons. What is more, the organization of the taste network in the brain would suggest that the parameters of space and time extend to the neural processing of gustatory information on a much grander scale.


Sujet(s)
Potentiels d'action/physiologie , Calicules gustatifs/physiologie , Goût/physiologie , Animaux , Humains , Réseau nerveux/physiologie , Voies nerveuses/physiologie , Activation chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...